localized treatment with oncolytic adenovirus delta-24 ......2019/08/27  · 1 localized treatment...

35
1 Localized Treatment with Oncolytic Adenovirus Delta-24-RGDOX Induces Systemic Immunity against Disseminated Subcutaneous and Intracranial Melanomas Hong Jiang 1 , Dong Ho Shin 1 , Teresa T. Nguyen 1 , Juan Fueyo 1 , Xuejun Fan 1 , Verlene Henry 1 , Caroline C. Carrillo 2 , Yanhua Yi 1 , Marta M. Alonso 3 , Tiara L. Collier 1 , Ying Yuan 4 , Frederick F. Lang 1 , and Candelaria Gomez-Manzano 1 1 Brain Tumor Center; 2 Applied Cancer Science Institute; 4 Department of Biostatistics; The University of Texas MD Anderson Cancer Center, Houston, Texas. 3 University Hospital of Navarra, Pamplona, Spain. Running Title: Delta-24-RGDOX induces systemic anti-melanoma immunity Keywords: oncolytic adenovirus, Delta-24-RGDOX, immunity, melanoma, metastasis Financial Support: This work was supported in part by NIH/NCI grants P50CA127001 and Cancer Center Support Grant P30CA016672 (Research Animal Support Facility, Small Animal Imaging Facility); the Cancer Prevention and Research Institute of Texas (RP170066); the Department of Defense (Team Science Award CA160525); the Marnie Rose Foundation; t he J.P. Harris Brain Tumor Research Fund; the Bradley Zankel Foundation; the Ruth L. Kirschstein National Research Service Award (NRSA) Individual Predoctoral Fellowship (Parent F31: 1 F31 CA228207 01A1), the American Legion Auxiliary Fellowship in Cancer Research; Terry and Janet Klebe Fellowship. Corresponding Authors: Hong Jiang, Department of Neuro-Oncology, Unit 1002, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030. Phone: 713-834-6203; Email: [email protected]. Research. on June 17, 2021. © 2019 American Association for Cancer clincancerres.aacrjournals.org Downloaded from Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 27, 2019; DOI: 10.1158/1078-0432.CCR-19-0405

Upload: others

Post on 01-Feb-2021

4 views

Category:

Documents


0 download

TRANSCRIPT

  • 1

    Localized Treatment with Oncolytic Adenovirus Delta-24-RGDOX Induces Systemic Immunity against

    Disseminated Subcutaneous and Intracranial Melanomas

    Hong Jiang1, Dong Ho Shin

    1, Teresa T. Nguyen

    1, Juan Fueyo

    1, Xuejun Fan

    1, Verlene Henry

    1, Caroline C.

    Carrillo2, Yanhua Yi

    1, Marta M. Alonso

    3, Tiara L. Collier

    1, Ying Yuan

    4, Frederick F. Lang

    1, and Candelaria

    Gomez-Manzano1

    1Brain Tumor Center;

    2Applied Cancer Science Institute;

    4Department of Biostatistics; The University of Texas

    MD Anderson Cancer Center, Houston, Texas. 3University Hospital of Navarra, Pamplona, Spain.

    Running Title: Delta-24-RGDOX induces systemic anti-melanoma immunity

    Keywords: oncolytic adenovirus, Delta-24-RGDOX, immunity, melanoma, metastasis

    Financial Support: This work was supported in part by NIH/NCI grants P50CA127001 and Cancer Center

    Support Grant P30CA016672 (Research Animal Support Facility, Small Animal Imaging Facility); the Cancer

    Prevention and Research Institute of Texas (RP170066); the Department of Defense (Team Science Award

    CA160525); the Marnie Rose Foundation; the J.P. Harris Brain Tumor Research Fund; the Bradley Zankel

    Foundation; the Ruth L. Kirschstein National Research Service Award (NRSA) Individual Predoctoral Fellowship (Parent

    F31: 1 F31 CA228207 01A1), the American Legion Auxiliary Fellowship in Cancer Research; Terry and Janet Klebe

    Fellowship.

    Corresponding Authors:

    Hong Jiang, Department of Neuro-Oncology, Unit 1002, The University of Texas MD Anderson Cancer Center,

    1515 Holcombe Blvd., Houston, TX 77030. Phone: 713-834-6203; Email: [email protected].

    Research. on June 17, 2021. © 2019 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 27, 2019; DOI: 10.1158/1078-0432.CCR-19-0405

    mailto:[email protected]://clincancerres.aacrjournals.org/

  • 2

    Candelaria Gomez-Manzano, Department of Neuro-Oncology, Unit 1002, The University of Texas MD

    Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030. Phone: 713-834-6221; Email:

    [email protected].

    Disclosure of Potential Conflicts of Interest: HJ, FFL, JF, and CGM are reporting ownership interest

    (including patents). CGM and JF are consultants and shareholders of DNATrix, Inc. . The authors have no

    other financial interests.

    Word Count: 4997

    Number of Figures: 6

    Research. on June 17, 2021. © 2019 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 27, 2019; DOI: 10.1158/1078-0432.CCR-19-0405

    mailto:[email protected]://clincancerres.aacrjournals.org/

  • 3

    Translational Relevance

    Immune checkpoint blockade revolutionized the therapy for melanoma. But the therapeutic benefit is

    limited to only a subset of patients with immunogenic (“hot”) tumors and is restrained by immune-related

    adverse events. We tested the effect of injecting oncolytic adenovirus Delta-24-RGDOX into primary

    subcutaneous melanoma on disseminated subcutaneous and intracranial melanomas in syngeneic mouse models.

    We observed systemic immune activation in the treated mice, resulting in the regression of both treated and

    untreated tumors. Thus, the virus is expected to be efficacious in patients with immunosuppressive (“cold”)

    tumors as well and, given its cancer-selective property and localized application, have better safety profile in

    cancer patients than immune checkpoint blockade. Importantly, this is the first report to demonstrate that

    localized treatment with an oncolytic virus in the subcutaneous tumor is able to reject intracranial tumors,

    suggesting Delta-24-RGDOX can be applied to patients with brain melanoma metastasis.

    Research. on June 17, 2021. © 2019 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 27, 2019; DOI: 10.1158/1078-0432.CCR-19-0405

    http://clincancerres.aacrjournals.org/

  • 4

    Abstract

    Purpose: Intratumoral injection of oncolytic adenovirus Delta-24-RGDOX induces efficacious anti-

    glioma immunity in syngeneic glioma mouse models. We hypothesized that localized treatment with the virus is

    effective against disseminated melanomas.

    Experimental Design: We tested the therapeutic effect of injecting Delta-24-RGDOX into primary

    subcutaneous (s.c.) B16-Red-FLuc tumors in s.c./s.c. and s.c./intracranial (i.c.) melanoma models in C57BL/6

    mice. Tumor growth and in vivo luciferase-expressing ovalbumin-specific (OT-I/Luc) T cells were monitored

    with bioluminescence imaging. Cells were profiled for surface markers with flow cytometry.

    Results: In both s.c./s.c. and s.c./i.c. models, 3 injections of Delta-24-RGDOX significantly inhibited

    the growth of both the virus-injected s.c. tumor and untreated distant s.c. and i.c. tumors, thereby prolonging

    survival. The surviving mice were protected from rechallenging with the same tumor cells. The virus treatment

    increased the presence of T cells and the frequency of effector T cells in the virus-injected tumor and mediated

    the same changes in T cells from peripheral blood, spleen, and brain hemispheres with untreated tumor.

    Moreover, Delta-24-RGDOX decreased the numbers of exhausted T cells and regulatory T cells in the virus-

    injected and untreated tumors. Consequently, the virus promoted the in situ expansion of tumor-specific T cells

    and their migration to tumors expressing the target antigen.

    Conclusions: Localized intratumoral injection of Delta-24-RGDOX induces an in situ antovaccination

    of the treated melanoma, the effect of which changes the immune landscape of the treated mice, resulting in

    systemic immunity against disseminated s.c. and i.c. tumors.

    Research. on June 17, 2021. © 2019 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 27, 2019; DOI: 10.1158/1078-0432.CCR-19-0405

    http://clincancerres.aacrjournals.org/

  • 5

    Introduction

    Because of its propensity to metastasize throughout the body, melanoma is a deadly tumor, accounting

    for the majority (60–80%) of deaths from skin cancer (1, 2). Ninety-five percent of melanoma patients with 3 or

    more sites of metastatic disease die within 1 year (3). Over the past decade, immune checkpoint blockade (ICB)

    has significantly improved the clinical outcomes of melanoma patients. However, the clinical benefit of ICB is

    preferentially achieved in only a subset of patients with a pre-existing T-cell response against the tumor (“hot”

    tumor) (4-8). Moreover, because checkpoint receptors play important regulatory roles in autoimmunity, the

    toxicity of ICB therapies causes immune-related adverse events (IRAEs). The IRAE incidence ranges from 70%

    in patients treated with αPD-1/αPD-L1 antibodies to as high as 90% in patients treated with αCTLA-4 (9).

    Therefore, safe and efficacious therapies are imperatively needed.

    Oncolytic viruses are a new class of immunotherapy drugs (10). They are genetically modified or

    naturally occurring viruses that selectively replicate in and disrupt cancer cells (11-13). In 2015, talimogene

    laherparepvec (T-VEC), a modified herpes simplex virus type 1 with cancer-selective replication and

    granulocyte-macrophage colony-stimulating factor (GM-CSF) expression, became the first oncolytic virus

    approved by U.S. Food and Drug Administration (FDA) for the treatment of surgically unresectable skin and

    lymph node lesions in patients with advanced melanoma (14, 15). In a randomized open-label phase III trial,

    those in the T-VEC arm achieved a better durable response rate than those in the control arm did (16% versus

    2%; median overall survival duration, 23.3 versus 18.9 months) (14). Thus, even though T-VEC has not

    dramatically improved melanoma therapy, it marks the beginning of a new era in which metastatic cancers can

    be treated with localized intratumoral injections of oncolytic viruses, which induced the regression of both the

    virus-injected and distant tumors, including visceral metastases (16).

    In a recent phase I clinical trial based on our long-term effort to develop oncolytic adenoviruses for

    cancer therapy, an intratumoral injection of Delta-24-RGD (DNX-2401, Tasadenoturev), a second-generation

    oncolytic adenovirus that is derived from human adenovirus serum type 5 and targets tumor cells with an

    aberrant RB pathway (17-19), elicited inflammation within the injected tumors, leading to a durable complete

    Research. on June 17, 2021. © 2019 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 27, 2019; DOI: 10.1158/1078-0432.CCR-19-0405

    http://clincancerres.aacrjournals.org/

  • 6

    response in 12% of patients with recurrent glioblastoma (20). To further increase the efficacy of the virus, we

    developed a next-generation oncolytic adenovirus, Delta-24-RGDOX, which expresses the immune stimulatory

    OX40 ligand (OX40L). Compared with its predecessor, this new virus elicits a more potent autologous cancer

    vaccination in situ, resulting in an efficacious, tumor-specific, long-lasting therapeutic effect in

    immunocompetent mouse glioma models (21). Because the virus preferentially expresses OX40L on the surface

    of infected tumor cells, thereby stimulating activated T cells that recognize the antigens presented on the tumor

    cells (21), Delta-24-RGDOX may be more cancer-specific and have a better safety profile in patients than T-

    VEC, which secrets GM-CSF in the tumor milieu (16). A clinical trial of Delta-24-RGDOX in glioma patients

    has been approved in Spain and is pending approval by the U.S. FDA.

    In our experience, Delta-24-RGD and Delta-24-RGDOX induce T-cell infiltration in the virus-injected

    glioma and immune memory against the treated tumor (20-22), suggesting that the viruses are able to turn

    immune-suppressive (“cold”) tumors into immune-active (“hot”) tumors. We hypothesized that Delta-24-

    RGDOX has the same effect in melanoma and that the virus-mediated therapeutic effect could extend to

    untreated disseminated/metastatic tumors. To test this hypothesis, we examined the virus in subcutaneous

    (s.c.)/s.c. and s.c./i.c. syngeneic melanoma models in immunocompetent mice. We found that injections of

    Delta-24-RGDOX not only induced immunity against the virus-injected s.c. tumor but also affected distant

    untreated tumors and changed the immune landscape in the treated mice. For the first time, we demonstrated

    this type of virotherapy induced efficacious immunity against melanoma in brain. Our findings suggest that

    localized treatment with Delta-24-RGDOX can be an effective therapeutic agent for patients with metastatic

    tumors, including brain metastasis.

    Materials and Methods

    Cell lines and culture conditions

    Human lung carcinoma A549 cells (ATCC) were cultured in Dulbecco’s modified Eagle’s medium

    nutrient mixture F12 (DMEM/F12) supplemented with 10% fetal bovine serum (FBS; HyClone Laboratories,

    Research. on June 17, 2021. © 2019 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 27, 2019; DOI: 10.1158/1078-0432.CCR-19-0405

    http://clincancerres.aacrjournals.org/

  • 7

    Inc.), 100 μg/ml penicillin, and 100 μg/ml streptomycin. The mouse melanoma cell lines B16-F10 (ATCC),

    B16F10 Red-FLuc (PerkinElmer), B16F10 Red-FLuc-3 (a subclone of B16F10 Red-FLuc with slower growing

    s.c. and i.c. tumors), and B16-ovalbumin (OVA; a kind gift from Dr. Hong Qin, MD Anderson Cancer Center)

    (23, 24) were grown in RPMI 1640 medium supplemented with 10% FBS and antibiotics. Human embryonic

    kidney 293 cells (QBioGene, Inc.) human melanoma A375 cells (ATCC) and mouse lung carcinoma CMT64

    cells (Culture Collections, Public Health England, UK) were maintained in DMEM supplemented with 10%

    FBS and antibiotics. All cells were kept at 37°C in a humidified atmosphere containing 5% CO2. Experiments

    were carried out within 6 months after the cell lines were obtained from a cell bank or within 15 passages in the

    laboratory. All cell lines were tested for and found to be free of mycoplasma.

    B16-F10enhanced green fluorescent protein (EGFP) cells

    First, EGFP cDNA (Clontech) was subcloned into the lentivector plasmid pCDH-CMV-MCS-EF2-Puro

    (System Biosciences). The plasmid was transduced into B16-F10 cells with X-tremeGENE HP DNA

    Transfection Reagent (Millipore Sigma), and a stable cell line was established by selection with 1 µg/ml

    puromycin.

    Viruses

    The Delta-24-RGD and Delta-24-RGDOX viruses were propagated in A549 cells and titrated in 293

    cells as described previously (21).

    Mice

    C57BL/6 mice were provided by MD Anderson’s Mouse Resource Facility. OT-I/Luc mice (a kind gift

    from Dr. J.J. Moon, University of Michigan) were bred at MD Anderson’s animal facility as described

    previously (25).

    Research. on June 17, 2021. © 2019 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 27, 2019; DOI: 10.1158/1078-0432.CCR-19-0405

    http://clincancerres.aacrjournals.org/

  • 8

    Animal studies

    Six- to ten-week-old mice were used. B16-F10 cells and their derivatives (5 × 105

    cells/mouse) were

    grafted subcutaneously onto the backs of the mice. B16F10 Red-Fluc-3 (2,000 cell/mouse) were grafted into the

    caudate nucleus of the mice using a guide-screw system as described previously (18). One day before virus

    treatment, the mice with implanted tumors were evaluated with bioluminescent imaging to assess tumor growth.

    Mice with off-size tumors were eliminated from the study, and the rest of the mice were randomly assigned to

    experimental groups. Delta-24-RGDOX (2 × 108

    plaque-forming units [PFUs]/mouse) was injected into the first

    implanted s.c. tumor. For rechallenging the surviving mice, B16F10 Red-FLuc (5 × 105

    cells /mouse) or CMT64

    (1× 106

    cells /mouse) cells were implanted s.c.; or B16F10 Red-FLuc-3 (2,000

    cells /mouse) cells were

    implanted in the contralateral hemisphere of the mouse brain. For monitoring tumor-specific T cells in vivo, 1×

    106

    OT-I/Luc CD8+ T cells /mouse were injected into the primary s.c. tumor one day before the virotherapy. In

    the s.c./s.c. model, mice were euthanized when the sum of the longer diameters of the two s.c. tumors was ≥ 2

    cm. In the s.c./i.c. model, mice were euthanized when either the longer diameter of s.c. tumor was ≥ 2 cm or the

    mice showed neurological disorders, such as hunched posture, loss of weight and slow movement. All animal

    studies were conducted in C57BL/6 mice. All experimental procedures involving the use of mice were done in

    accordance with protocols approved by MD Anderson’s Animal Care and Use Committee and followed U.S.

    National Institutes of Health and Department of Agriculture guidelines.

    Bioluminescence imaging of tumor-bearing mice

    Following implantation of the bioluminescent cells, the luminescence from the growing tumors in the

    mice was detected with the IVIS Spectrum imaging system (PerkinElmer). Briefly, each mouse was injected

    subcutaneously with 200 μl D-luciferin (Gold Biotechnology; 15 mg/ml in phosphate-buffered saline [PBS]).

    Ten minutes later, the animals were anesthetized by placing them in a chamber with 2% isoflurane gas in O2

    until they were unresponsive. The anesthetized animals were moved to the imaging chamber to acquire images

    Research. on June 17, 2021. © 2019 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 27, 2019; DOI: 10.1158/1078-0432.CCR-19-0405

    http://clincancerres.aacrjournals.org/

  • 9

    of the tumors. The luminescence in the mice was assessed with the Living Image software program

    (PerkinElmer).

    Immunofluorescence staining

    Paraffin-embedded tumor tissue was cut into 5-µm sections and processed for immunofluorescence

    staining with standard procedures. The primary antibodies were anti-E1A (MA1-91861; Invitrogen); the

    secondary antibodies were anti-mouse IgG Alexa Fluor (A11001). Finally, the slides were mounted with cover

    slips using a ProLong Gold antifade reagent (Invitrogen) containing DAPI for nuclei staining. Images were

    viewed and assessed using a FluoView confocal laser scanning microscope (Olympus).

    Enrichment of leukocytes from tumors

    The enrichment of leukocytes from s.c. tumors was conducted as described previously with modification

    (26). Briefly, the tumors were collected, placed in a 100-μm cell strainer set in petri dishes with RPMI 1640

    medium, and then processed through the cell strainer into the dish. The mixture in the dish was gently pipetted

    up and down and brought up to 20 ml/g of tumor. After centrifugation for 5 min at 500×g, the pellet was

    resuspended in 10 ml of medium and loaded on top of a discontinuous Percoll gradient medium (44%:67%).

    After centrifugation at 800×g for 30 minutes at 4°C, the cells were retrieved from the 44%-67% interphase.

    Brain-infiltrating leukocytes from mouse brain hemispheres were separated from myelin debris using

    Percoll gradient medium (GE Healthcare Bio-Sciences) and centrifuged as described previously (27).

    Preparation of splenocytes, lymph node cells, and CD8+ T lymphocytes

    Mouse spleens and lymph nodes were collected and processed as described previously, except that the

    lymph node cells were not subjected to red blood cell lysis (21). CD8+ T cells were enriched from the

    splenocytes with a mouse CD8a+ T Cell Isolation Kit (Miltenyi Biotec, Inc.).

    Research. on June 17, 2021. © 2019 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 27, 2019; DOI: 10.1158/1078-0432.CCR-19-0405

    http://clincancerres.aacrjournals.org/

  • 10

    IFNγ ELISPOT assay

    The ELISPOT assay was performed with Mouse IFNγ ELISpot kit from R&D systems. Briefly,

    splenocytes (5 × 105 per well) were stimulated with 1 µg/ ml CD8+ T cell-specific OVA (257-264) (Invitrogen)

    or GFP (118-126) peptide (Biomatik) for 21 hrs. IFNγ spot-forming cells were determined according to the

    instructions from the manufacturer.

    Flow cytometry analysis

    To analyze cell surface protein expression, we incubated cells (2-5 × 105) in 100 µl of primary antibody

    solution diluted in PBS plus 3% bovine serum albumin and 1 mM ethylenediaminetetraacetic acid (EDTA).

    After incubation at 4°C in the dark for 30 minutes, the cells were washed once with 1 ml of cold PBS. Then, the

    cells were resuspended in 0.5 ml of PBS. The stained cells were analyzed using flow cytometry. The antibodies

    used in the studies were to the following mouse proteins: CD252 (OX40L) allophycocyanin (APC; 17-5905-

    80), CD3e phycoerythrin (PE; 12-0031-81), CD4 APC (17-0041-81), CD8a PE-Cyanine5.5 (35-0081-80), PD-1

    fluorescein isothiocyanate (11-9981-81), TIM3 PE-Cyanine7 (25-5870-80), CD44 super bright 600 (63-0441-

    80), CD62L APC-eFlour 780 (47-0621-80), CD25 PE-Cyanine5.5 (35-0251-80), FR4 PE-Cyanine7 (25-5445-

    80), MHC Class I (H-2Kb) APC (17-5958-80 ), Mouse IgG2a kappa Isotype Control APC (17-4724-81), Rat

    IgG2b kappa Isotype Control (14-4031-82), MHC Class II (I-A/I-E, 14-5321-81), from eBioscience; and CD45

    BV786 (564225), from BD Biosciences. The goat anti-rat IgG F(ab')2-APC (sc-3832) was from Santa Cruz

    Biotechnology, Inc.. Tetramer/PE - H-2 Kb OVA (SIINFEKL) (OVA-Tet; 14087-PE Kb/OVA) was from the

    MHC Tetramer Production Facility, Baylor College of Medicine. Cell counts were obtained using 123count

    eBeads (01-1234, eBioscience) as a control for calculating processed sample volumes. Dead cells were

    excluded from the analysis through staining with Ghost Dye Violet 510 (13-0870, Tonbo Biosciences).

    Stimulation of immune cells

    Research. on June 17, 2021. © 2019 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 27, 2019; DOI: 10.1158/1078-0432.CCR-19-0405

    http://clincancerres.aacrjournals.org/

  • 11

    For preparation of the target cells, B16F10 Red-FLuc cells were infected with the virus (100 PFU/cell).

    Four hours later, 50 units/ml mouse interferon gamma (IFNγ; Prospec Protein Specialists) was added to the

    culture. Forty-eight hours after viral infection, the cells were fixed with 1% paraformaldehyde and washed

    before added to the co-culture. For immune cell stimulation, pre-fixed target cells (2 × 104 cells/well)

    were

    incubated with splenocytes or lymph node cells (5 × 105 cells/well). Forty hours after the co-culture in a round-

    bottom 96-well plate, the concentration of IFNγ in the medium was assessed with a Mouse IFNγ DuoSet ELISA

    kit (R&D Systems).

    Statistical analysis

    In quantitative studies of cultured cells and flow cytometry, each group consisted of triplicate samples.

    Each study was repeated at least once. Differences between groups were evaluated using a 2-tailed Student t-

    test. The animal survival curves were plotted according to the Kaplan–Meier method. Survival rates in the

    different treatment groups were compared using the log-rank test. Waterfall charts were plotted with R from

    http://www.R-project.org. P values < 0.05 were considered significant.

    Results

    Localized treatment with Delta-24-RGDOX inhibits treated primary and untreated distant s.c.

    melanomas and induces immune memory

    It has been reported that oncolytic adenoviruses have shown oncolytic efficacy in melanomas (28, 29).

    Thus, first we tested Delta-24-RGDOX in cultured human and mouse melanoma cells. The virus infected both

    human and mouse melanoma cells and expressed OX40L efficiently in these cells (Supplementary Fig. S1A).

    The B16F10 and its derivatives used in our studies are comparably sensitive to Delta-24-RGDOX infection. 48

    hours after viral infection at 30 pfu/cell, the percentage of OX40L positive cells ranged from 50% to 96%

    among these cell lines (Supplementary Fig. S1B). Three days after the virus was injected into a s.c. melanoma

    derived from B16F10-EGFP cells, OX40L expression was detected in about 11% of the tumor cells but not in

    Research. on June 17, 2021. © 2019 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 27, 2019; DOI: 10.1158/1078-0432.CCR-19-0405

    http://clincancerres.aacrjournals.org/

  • 12

    the PBS-treated tumor (P = 0.0002; Supplementary Fig. S1C). This virus replicated in human melanoma A375

    cells, albeit not as efficiently as in human lung cancer A549 cells, which are optimal hosts for adenovirus

    replication. The replication of Delta-24-RGDOX was detectable in mouse melanoma B16F10 Red-FLuc cells

    (Supplementary Fig. S1D) and the virus lysed the cells efficiently (Supplementary Fig. S1E).

    Next, we examined the anti-melanoma activity of the virus in an s.c./s.c. syngeneic melanoma model,

    derived from B16F10 Red-FLuc cells, in immunocompetent C57BL/6 mice (Fig. 1A). Monitoring of tumor

    growth with bioluminescence imaging ((Fig. 1B) revealed that 3 injections of Delta-24-RGDOX into the

    primary s.c. tumor inhibited both the treated tumor and untreated distant tumors (Fig. 1C, Supplementary Fig.

    S2), thereby prolonging survival and resulting in a long-term survival rate of 44% in the tumor-bearing mice

    (median survival durations: 36 vs 16 days, P = 0.005, Fig. 1D). The survivors of the virus treatment were

    protected from rechallenging with s.c. injection of the same tumor cells but not CMT64 cells of lung carcinoma

    (Fig. 1E), indicating that the treatment induced immune memory specifically against the virus-treated tumor.

    Localized treatment with Delta-24-RGDOX inhibits treated s.c. primary and untreated distant i.c.

    melanomas

    Consistently, in the s.c./i.c. melanoma model derived from B16F10 Red-FLuc-3 cells, a subclone of

    B16F10 Red-FLuc cells that grow slower than the parental cells in the i.c. tumor and give enough window

    period for us to evaluate the effect of Delta-24-RGDOX in this model (Fig. 2A), viral injections into the

    primary s.c. tumor inhibited not only the treated tumor but also the untreated i.c. tumor (Fig. 2B and C,

    Supplementary Fig. S3), thereby prolonging survival and resulting in a long-term survival rate of 40% in the

    tumor-bearing mice (median survival durations: 58 vs 27 days, P = 0.005, Fig. 2D). Since the s.c. tumor grew

    faster, all the mice in PBS group were euthanized because of s.c. tumor burden and 6 of the brains showed

    visible tumor. Because the virus inhibited s.c. tumor growth, in the D24-RGDOX group, of the six euthanized

    mice, 4 died of brain tumor, 2 died of s.c. tumor. The survivors of the virus treatment were resistant to

    rechallenging with the same type of tumor cells in the contralateral hemisphere of the brain (Fig. 2E and F),

    Research. on June 17, 2021. © 2019 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 27, 2019; DOI: 10.1158/1078-0432.CCR-19-0405

    http://clincancerres.aacrjournals.org/

  • 13

    indicating that the treatment induced immune memory against the virus-treated tumor. Moreover, we found no

    pathological changes in the brains from the survivors (Figure 2F, lower panel).

    Collectively, these data demonstrate that the anti-cancer effect of intratumoral injection of Delta-24-

    RGDOX could extend from the treated tumor to the distant untreated s.c. tumor and could even pass the blood-

    brain barrier and reach the brain tumor, meaning that localized treatment of a primary s.c. tumor with the virus

    can have a efficacious therapeutic effect on disseminated/metastatic tumors.

    Intratumoral injection of Delta-24-RGDOX increases the presence of T cells and activates these cells at

    the virus-injected tumor

    To study the effect induced by Delta-24RGDOX within the injected tumor, we first checked the tumor

    24 h after viral injection. Immunostaining revealed that the virus expressed the early protein E1A in the infected

    tumor cells at this time (Fig. 3A). Two days after the viral injections, we also observed virus-mediated

    upregulation of MHC I (about 6-fold increase) and MHC II (almost doubled) expression on melanoma cells

    (Fig. 3B), indicating the tumor cells are more active in presenting tumor-associated antigens to the immune

    cells. Three days after the viral injections, the number of leukocytes (CD45+ cells) nearly doubled in the treated

    tumor (Fig. 3C). The frequency of T cells (CD45+ CD3+) tripled in the virus-treated tumors (from 20.7% to

    63.4%), while the frequency of cytotoxic T cells (CD8+) increased by 23% (P < 0.0001) and that of T helper

    cells (CD4+) showed little change (P = 0.9; Fig. 3D). However, owing to the increase of the leukocytes in the

    tumor, the numbers of T cells, cytotoxic T cells, and helper T cells all increased dramatically in the tumors (Fig.

    3E). Further analysis of the T-cell populations revealed that the virus activated T cells, thereby increasing the

    frequency and number of effector (CD44+ CD62L-) CD8+ or CD4+ T cells within the virus-treated tumor (Fig.

    3F). Thus, intratumoral injection of Delta-24-RGDOX caused immune activation in the tumor.

    Localized treatment of the tumor with Delta-24-RGDOX causes systemic activation of T cells in tumor-

    bearing mice

    Research. on June 17, 2021. © 2019 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 27, 2019; DOI: 10.1158/1078-0432.CCR-19-0405

    http://clincancerres.aacrjournals.org/

  • 14

    Since the injection of Delta-24-RGDOX in the primary s.c. tumor mediated the regression of both the

    treated tumor and disseminated untreated tumors (Fig. 1 and 2), we hypothesized that the virus could change the

    entire immune landscape in the treated mice while inducing the same effect in untreated tumors. Indeed, we

    found that in peripheral blood, the frequency of T cells and cytotoxic T cells increased after virus treatment (P ≤

    0.001), whereas the frequency of T helper cells declined slightly (P = 0.02; Fig. 4A). The frequency of T

    effector CD8+ T cells almost doubled (from 12.9% to 25.1%), whereas that of CD4+ T cells barely changed (P

    = 0.08; Fig. 4B). Consistent with these findings, in the hemispheres with implanted tumors, Delta-24-RGDOX

    induced the following changes: the frequency of T cells more than doubled after virus injections in the s.c.

    tumor; the frequency of CD8+ T cells increased by 28% (P = 0.0001), whereas the frequency of CD4+ T cells

    remained almost the same (P = 0.1; Fig. 4C); the numbers of total T cells, CD4+ T cells, and CD8+ T cells

    increased by 85%137% (Supplementary Fig. S4); and the numbers of effector T cells in both the CD8+ and

    CD4+ T-cell populations nearly doubled (Fig. 4D). As in peripheral blood, after virus treatment, the frequency

    of effector CD8+ T cells increased 2-fold in splenocytes and increased by 60% in cells from inguinal lymph

    nodes (the tumor-draining lymph nodes [TDLNs] for s.c. tumors), whereas the frequency of CD4+ T cells

    increased only slightly in splenocytes and declined in TDLN cells (Supplementary Fig. S5). When splenocytes

    or inguinal lymph node cells were co-cultured with melanoma cells with or without viral infection, by assessing

    the IFNγ secretion into the medium, we found that the immune cells from the virus-treated group demonstrated

    higher activity against the tumor cells (Fig. 4E). Furthermore, Delta-24-RGDOXinfected melanoma cells

    stimulated immune cell activity more potently than Delta-24-RGDinfected cells or cells without viral infection

    did (Fig. 4E), suggesting that OX40L expression on the cell surface enhances the capability of tumor cells to

    induce anti-cancer immunity, which is consistent with what we observed previously in glioma virotherapy (21).

    Delta-24-RGDOX disrupts immune suppression in both treated primary and untreated distant tumors

    To better understand the effect of virotherapy on immunity, we further examined the status of exhausted

    T cells (PD-1+ TIM3+) (30) and regulatory T cells (CD4+ CD25+ FR4+) (31). First, we analyzed these cell

    Research. on June 17, 2021. © 2019 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 27, 2019; DOI: 10.1158/1078-0432.CCR-19-0405

    http://clincancerres.aacrjournals.org/

  • 15

    populations in the tumors. We found that Delta-24-RGDOX reduced the frequency of exhausted CD8+ T cells

    in both virus-injected s.c. tumors (from 37% to 10.5%, P = 0.000001) and untreated i.c. tumors (from 30.1% to

    14.4%, P = 0.001; Fig. 5A). The frequency of exhausted CD4+ T cells was also reduced in these 2 tumor

    locations, albeit to a lesser extent (P < 0.005; Fig. 5A). As reported previously (30), all the TIM3+ T cells were

    PD-1+ (Fig. 5A). Like its effect on T-cell exhaustion, Delta-24-RGDOX decreased the frequency of regulatory

    T cells in both the virus-injected s.c. tumors (from 20.3% to 5.6%, P = 0.00004) and untreated i.c. tumors (from

    30.7% to 14.0%, P = 0.00001) (Fig. 5B). However, exhausted T cells were barely detectable in peripheral blood

    and inguinal lymph nodes (Supplementary Fig. S6A), and regulatory T-cell frequency was hardly detected in

    blood (Supplementary Fig. S4B, left panel). Although we observed a regulatory T-cell frequency of 89% in

    inguinal lymph nodes, the virus treatment had little effect on the frequency of this cell population in the tissue

    (Supplementary Fig. S6B, right panel). Interestingly, in all the tissues we examined, we observed the virus-

    mediated increase of PD-1+ cytotoxic T cell frequency with the highest in untreated i.c. tumors (Fig. 5C).

    Intratumoral injection of Delta-24RGDOX mediates in situ tumor-specific T-cell expansion and

    migration to distant untreated tumors

    To examine the relevance of the virus-mediated effect on immunity to tumor-specific T-cell response,

    we used the OVA protein as a model tumor-associated antigen. We set up the s.c./s.c. tumors as depicted in

    Figure 6A. Six days after the implantation of the first tumor and 1 day after the implantation of the second

    tumor, OVA-specific CD8+ T cells with luciferase expression (OT-I/Luc T cells) were injected into the first

    tumor, followed by 3 doses of viral injections in the same tumor. As demonstrated by bioluminescence imaging

    of OT-I/Luc T cells (Fig. 6B), Delta-24-RGDOX treatment promoted the migration of this cell population to

    distant OVA+ tumors but not OVA- tumors 4 days after the last dose of viral injection (Fig. 6C). Consistent

    with these findings, 3 days later, quantification of OT-I/Luc T cells in the tumors with flow cytometry analysis

    revealed that Delta-24-RGDOX treatment increased the number of these cells in the first injected tumor and

    greatly enhanced the presence of the cells in the second OVA+ tumors but not OVA- tumors (Fig. 6D),

    Research. on June 17, 2021. © 2019 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 27, 2019; DOI: 10.1158/1078-0432.CCR-19-0405

    http://clincancerres.aacrjournals.org/

  • 16

    suggesting that the virus can activate tumor-specific cytotoxic T cells and increase their in situ presence at the

    treated tumor and their migration to distant tumors expressing the T-cell target in this artificial antigen system.

    Consistently, Delta-24-RGDOX treatment mediated the increase of OVA-specific CD8+ T cells frequency in

    the spleens of the mice bearing s.c. tumors derived from B16-OVA cells (Fig. 6E).

    Discussion

    Our findings demonstrate that localized treatment with Delta-24-RGDOX induces immunity against

    both virus-injected tumors and untreated distant tumors in syngeneic s.c./s.c. and s.c./i.c. melanoma mouse

    models. The viral injections into the first s.c. tumor immunologically activate the tumor microenvironment of

    not only the treated tumor but also the distant untreated tumors, resulting in the regression of both the treated

    primary s.c. tumor and disseminated untreated s.c. and i.c. tumors. In addition, the virus also causes immune

    activation in peripheral blood and lymphoid organs. Importantly, the virus treatment stimulates in situ tumor-

    specific cytotoxic T-cell expansion and migration to distant untreated tumors expressing the T-cell target.

    Based on the evidence from preclinical and clinical experience, unlike ICB, oncolytic virotherapies

    should be effective in patients with “cold” tumors. Moreover, localized treatment of the tumor with cancer-

    selective oncolytic viruses is expected to cause fewer IRAEs than global immune checkpoint inhibition. In the

    phase I clinical trial of Delta-24-RGD in patients with recurrent malignant glioma, patients tolerated intratumal

    injection of the virus (up to 3 × 1010

    viral particles) well (20). Further, we reported previously that, through

    expressing immune stimulator OX40L on cancer cells, Delta-24-RGDOX was more potent than Delta-24-RGD

    in inducing tumor-specific immunity and immune memory against gliomas in orthotopic mouse models (21).

    During these studies, we observed no virotherapy toxicities, such as weight loss or neurological disorders, in

    long-term surviving mice. In our current study of virotherapy with Delta-24-RGDOX in melanoma mouse

    models, in the long-term surviving mice, we also found no sign of treatment-related toxicity, such as weight

    loss, neurological disorders, or chronic inflammation indicated by swelling and redness at the injection site.

    These observations suggest that Delta-24-RGDOX is well-tolerated in tumor-bearing mice.

    Research. on June 17, 2021. © 2019 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 27, 2019; DOI: 10.1158/1078-0432.CCR-19-0405

    http://clincancerres.aacrjournals.org/

  • 17

    Tumors escape immune surveillance through inactivating tumor-specific T cells in the tissue

    microenvironment, posing obstacles in cancer immunotherapy (32). T cells are inactivated through multiple

    tumor-intrinsic and -extrinsic immunosuppressive mechanisms (33). One such mechanism is tumor cells’

    expression of PD-L1, which ligates to immune checkpoint receptor PD-1 on the surfaces of activated T cells,

    natural killer cells, macrophages, and several subsets of dendritic cells, thereby impairing T-cell receptor (TCR)

    signaling and CD28 co-stimulation (34-36). Upon TCR activation, PD-1 expression is transiently induced on

    naïve T cells, and this expression decreases in the absence of TCR signaling but is maintained by chronic

    activation with a persisting epitope target (34). We observed much higher PD-1 expression on CD8+ T cells

    from tumors than on T cells from peripheral blood and lymphoid organs (Fig. 5C), which suggests that T cells

    have a feedback response to continuous stimulation by tumor antigens within the tumor. As in glioma mouse

    models described previously (21), PD-L1 was highly expressed on cultured melanoma cells with or without

    Delta-24-RGDOX infection and was greatly upregulated by IFNγ which is a pro-inflammatory cytokine

    induced by oncolytic adenovirus in vivo (22) (Supplementary Fig. S7A). Through their ligation to PD-1 on T

    cells, these cells stay at an anergy status and allow the immune escape of tumor cells. Delta-24-RGDOX

    treatment increased the PD-L1 expression level of melanoma cells from the treated tumors in mice

    (Supplementary Fig. S7B) and the upregulation of PD-1 expression on CD8+ T cells from treated and untreated

    tumors, peripheral blood, and lymphoid organs (Fig. 5C). Nonetheless, the virus treatment increased the activity

    of the immune cells against the tumor cells (Fig. 4E) and mediated the regression of both the treated and

    untreated tumors (Fig. 1 and 2), suggesting that PD-1 upregulation alone is not sufficient to inhibit T-cell

    activity. Actually, accumulating evidence indicates that PD-1 expression is an early marker of T-cell activation,

    allowing the identification of the tumor-reactive CD8+ T-cell fraction in melanoma tumors (37-39). T-cell

    inactivation needs the collaboration of PD-1 and other immune checkpoint inhibitors, such as TIM3 and TIGIT

    (40, 41). Consistent with the immune activation by the virus, in the present study, Delta-24-RGDOX decreased

    the frequency of PD-1+ TIM3+ T cells in treated and untreated tumors despite the increase in PD-1+ T cell

    frequency (Fig 5A and C). One study of PD-1targeted therapy in lung cancer patients revealed that the

    Research. on June 17, 2021. © 2019 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 27, 2019; DOI: 10.1158/1078-0432.CCR-19-0405

    http://clincancerres.aacrjournals.org/

  • 18

    upregulation of PD-1 expression on CD8+ T cells from peripheral blood within 4 weeks of treatment initiation

    correlates with clinical benefit (42). Thus, the early Delta-24-RGDOXinduced upregulation of PD-1 on CD8+

    T cells from peripheral blood might serve as a biomarker to predict patients' responses to the virotherapy. In

    addition, regulatory T cells selectively accumulate in the tumor microenvironment, suppressing T cell immune

    responses and activities of antigen-presenting cells, including DCs and macrophages (43). Consistent with the

    role of OX40 in inhibiting Foxp3 expression and regulatory T cell induction (44), Delta-24-RGDOX

    downregulated the frequency of regulatory T cells in treated and untreated tumors (Fig. 5B), which contributed

    to the immune activation within the tumors (Figs. 3, 4C and D).

    Melanoma has the potential to metastasize to virtually any anatomical site. Compared with other types

    of cancers, melanoma is more likely to metastasize to the brain, and brain metastasis occurs in more than 50%

    of patients with advanced disease (45). The median survival time after the detection of brain metastasis is 17–22

    weeks (45). The prognosis for patients with melanoma brain metastasis is dismal despite current chemotherapy,

    surgery, and radiation options (46). The blood-brain barrier (BBB) limits systemic chemotherapies’ penetration

    of the central nervous system (CNS), and the negative side effects of radiotherapy pose challenges to

    therapeutic success, and some lesions are not accessible for surgery (46). As with T-VEC virotherapy in

    melanoma patients (16), we observed that intratumoral injection of Delta-24-RGDOX into the first s.c. tumor

    led to immune activation in distant untreated melanomas (Fig. 4C and D), resulting in the regression of both

    treated and untreated i.c. tumors (Fig. 2). Meanwhile, we found T-cell activation in the peripheral blood, spleen,

    and inguinal lymph nodes after injection of Delta-24-RGDOX into the first s.c. tumor (Fig. 4A, B, and E).

    During brain metastasis, the BBB is selectively disrupted (47). In addition, recent studies indicate that, instead

    of being immune-privileged, the CNS actively communicates with the immune system through the lymphatic

    vascular system (48, 49). These contribute to the interaction between the CNS and immune system. Thus, the

    immune factors induced by localized treatment with Delta-24-RGDOX, including pro-inflammatory cytokines

    and immune cells, may reach the i.c. melanoma through BBB leakage or lymphatic circulation. In our study, the

    Research. on June 17, 2021. © 2019 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 27, 2019; DOI: 10.1158/1078-0432.CCR-19-0405

    http://clincancerres.aacrjournals.org/

  • 19

    anti-tumor immunity mediated by localized virus administration is strong enough to have an abscopal anti-

    melanoma effect in the brain (Fig. 2).

    Since mouse cells are not optimal host for human adenovirus type 5 replication (50), our current models

    have limitations to recapitulate the oncolytic effect of the virus in human melanomas although we detected low

    level Delta-24-RGDOX replication in B16F10 Red-FLuc cells (Supplementary Fig. S1D). We expect the virus

    will be more potent to replicate in human melanomas to initiate tumor destruction cascade and consequential

    anti-melanoma immunity. In addition, although the work has been done in tumor models from B16F10 and its

    derivative cell lines, we believe the virus should be able to induce similar effect in other solid tumors. We have

    shown previously this virus is efficacious to inhibit gliomas in mouse (21).

    In summary, we report for the first time that oncolytic adenovirus Delta-24-RGDOX effectively inhibits

    both treated primary s.c. and untreated distant s.c. and i.c. tumors, stimulates systemic immune activation in the

    treated mice, and promotes tumor-specific cytotoxic T-cell expansion and their targeting of disseminated

    untreated tumors. Our data provide strong evidence that this strategy can be further developed into an

    efficacious and safe clinical intervention for patients with advanced melanomas, including those with brain

    metastasis.

    Authors’ Contributions

    Conception and design: H. Jiang, J. Fueyo, C. Gomez-Manzano

    Development of methodology: H. Jiang

    Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): H. Jiang

    D. H. Shin, T. T. Nguyen, X. Fan, V. Henry, C. Carrillo, Y. Yi, T. L. Collier

    Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): H.

    Jiang, Y. Yuan

    Writing, review, and/or revision of the manuscript: H. Jiang, J. Fueyo, C. Gomez-Manzano, M. M. Alonso

    Research. on June 17, 2021. © 2019 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 27, 2019; DOI: 10.1158/1078-0432.CCR-19-0405

    http://clincancerres.aacrjournals.org/

  • 20

    Administrative, technical, or material support (i.e., reporting or organizing data, constructing

    databases): F. F. Lang

    Study supervision: H. Jiang, J. Fueyo, C. Gomez-Manzano

    Acknowledgements

    We thank Joe Munch (the Department of Scientific Publications, MD Anderson Cancer Center) for editing the

    manuscript.References

    1. Damsky WE, Theodosakis N, Bosenberg M. Melanoma metastasis: new concepts and evolving paradigms.

    Oncogene 2014;33: 2413-22.

    2. Sadozai H, Gruber T, Hunger RE, Schenk M. Recent Successes and Future Directions in Immunotherapy of

    Cutaneous Melanoma. Front Immunol 2017;8: 1617.

    3. Balch CM, Gershenwald JE, Soong SJ, Thompson JF, Atkins MB, Byrd DR, et al. Final version of 2009 AJCC

    melanoma staging and classification. J Clin Oncol 2009;27: 6199-206.

    4. Spranger S, Bao R, Gajewski TF. Melanoma-intrinsic beta-catenin signalling prevents anti-tumour immunity.

    Nature 2015;523: 231-5.

    5. Sharma PAllison JP. The future of immune checkpoint therapy. Science 2015;348: 56-61.

    6. Postow MA, Callahan MK, Wolchok JD. Immune Checkpoint Blockade in Cancer Therapy. J Clin Oncol 2015;33:

    1974-1982.

    7. Topalian SL, Drake CG, Pardoll DM. Immune checkpoint blockade: a common denominator approach to cancer

    therapy. Cancer Cell 2015;27: 450-61.

    8. Johnson DBSosman JA. Therapeutic Advances and Treatment Options in Metastatic Melanoma. JAMA Oncol

    2015;1: 380-6.

    9. Michot JM, Bigenwald C, Champiat S, Collins M, Carbonnel F, Postel-Vinay S, et al. Immune-related adverse

    events with immune checkpoint blockade: a comprehensive review. Eur J Cancer 2016;54: 139-148.

    Research. on June 17, 2021. © 2019 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 27, 2019; DOI: 10.1158/1078-0432.CCR-19-0405

    http://clincancerres.aacrjournals.org/

  • 21

    10. Kaufman HL, Kohlhapp FJ, Zloza A. Oncolytic viruses: a new class of immunotherapy drugs. Nat Rev Drug Discov

    2015;14: 642-62.

    11. Lichty BD, Breitbach CJ, Stojdl DF, Bell JC. Going viral with cancer immunotherapy. Nat Rev Cancer 2014;14: 559-

    67.

    12. Jiang H, Gomez-Manzano C, Rivera-Molina Y, Lang FF, Conrad CA, Fueyo J. Oncolytic adenovirus research

    evolution: from cell-cycle checkpoints to immune checkpoints. Curr Opin Virol 2015;13: 33-39.

    13. Russell SJ, Peng KW, Bell JC. Oncolytic virotherapy. Nat Biotechnol 2012;30: 658-70.

    14. Andtbacka RH, Kaufman HL, Collichio F, Amatruda T, Senzer N, Chesney J, et al. Talimogene Laherparepvec

    Improves Durable Response Rate in Patients With Advanced Melanoma. J Clin Oncol 2015;33: 2780-8.

    15. Sheridan C. First oncolytic virus edges towards approval in surprise vote. Nat Biotechnol 2015;33: 569-70.

    16. Conry RM, Westbrook B, McKee S, Norwood TG. Talimogene laherparepvec: First in class oncolytic virotherapy.

    Hum Vaccin Immunother 2018;14: 839-846.

    17. Fueyo J, Gomez-Manzano C, Alemany R, Lee PS, McDonnell TJ, Mitlianga P, et al. A mutant oncolytic adenovirus

    targeting the Rb pathway produces anti-glioma effect in vivo. Oncogene 2000;19: 2-12.

    18. Fueyo J, Alemany R, Gomez-Manzano C, Fuller GN, Khan A, Conrad CA, et al. Preclinical characterization of the

    antiglioma activity of a tropism-enhanced adenovirus targeted to the retinoblastoma pathway. J Natl Cancer Inst

    2003;95: 652-60.

    19. Jiang H, Gomez-Manzano C, Aoki H, Alonso MM, Kondo S, McCormick F, et al. Examination of the therapeutic

    potential of Delta-24-RGD in brain tumor stem cells: role of autophagic cell death. J Natl Cancer Inst 2007;99:

    1410-4.

    20. Lang FF, Conrad C, Gomez-Manzano C, Yung WKA, Sawaya R, Weinberg JS, et al. Phase I Study of DNX-2401

    (Delta-24-RGD) Oncolytic Adenovirus: Replication and Immunotherapeutic Effects in Recurrent Malignant

    Glioma. J Clin Oncol 2018;36: 1419-1427.

    21. Jiang H, Rivera-Molina Y, Gomez-Manzano C, Clise-Dwyer K, Bover L, Vence LM, et al. Oncolytic Adenovirus and

    Tumor-Targeting Immune Modulatory Therapy Improve Autologous Cancer Vaccination. Cancer Res 2017;77:

    3894-3907.

    Research. on June 17, 2021. © 2019 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 27, 2019; DOI: 10.1158/1078-0432.CCR-19-0405

    http://clincancerres.aacrjournals.org/

  • 22

    22. Jiang H, Clise-Dwyer K, Ruisaard KE, Fan X, Tian W, Gumin J, et al. Delta-24-RGD oncolytic adenovirus elicits anti-

    glioma immunity in an immunocompetent mouse model. PLoS One 2014;9: e97407.

    23. Falo LD, Jr., Kovacsovics-Bankowski M, Thompson K, Rock KL. Targeting antigen into the phagocytic pathway in

    vivo induces protective tumour immunity. Nat Med 1995;1: 649-53.

    24. Park HJ, Qin H, Cha SC, Sharma R, Chung Y, Schluns KS, et al. Induction of TLR4-dependent CD8+ T cell immunity

    by murine beta-defensin2 fusion protein vaccines. Vaccine 2011;29: 3476-82.

    25. Ochyl LJMoon JJ. Whole-animal imaging and flow cytometric techniques for analysis of antigen-specific CD8+ T

    cell responses after nanoparticle vaccination. J Vis Exp 2015: e52771.

    26. Rapp M, Anz D, Schnurr M. Isolation of intratumoral leukocytes of TLR-stimulated tumor-bearing mice. Methods

    Mol Biol 2014;1169: 175-9.

    27. LaFrance-Corey RGHowe CL. Isolation of brain-infiltrating leukocytes. J Vis Exp 2011: e2747.

    28. Schipper H, Alla V, Meier C, Nettelbeck DM, Herchenroder O, Putzer BM. Eradication of metastatic melanoma

    through cooperative expression of RNA-based HDAC1 inhibitor and p73 by oncolytic adenovirus. Oncotarget

    2014;5: 5893-907.

    29. Rodriguez-Garcia A, Gimenez-Alejandre M, Rojas JJ, Moreno R, Bazan-Peregrino M, Cascallo M, et al. Safety and

    efficacy of VCN-01, an oncolytic adenovirus combining fiber HSG-binding domain replacement with RGD and

    hyaluronidase expression. Clin Cancer Res 2015;21: 1406-18.

    30. Sakuishi K, Apetoh L, Sullivan JM, Blazar BR, Kuchroo VK, Anderson AC. Targeting Tim-3 and PD-1 pathways to

    reverse T cell exhaustion and restore anti-tumor immunity. J Exp Med 2010;207: 2187-94.

    31. Yamaguchi T, Hirota K, Nagahama K, Ohkawa K, Takahashi T, Nomura T, et al. Control of immune responses by

    antigen-specific regulatory T cells expressing the folate receptor. Immunity 2007;27: 145-59.

    32. Anderson KG, Stromnes IM, Greenberg PD. Obstacles Posed by the Tumor Microenvironment to T cell Activity: A

    Case for Synergistic Therapies. Cancer Cell 2017;31: 311-325.

    33. Pitt JM, Vetizou M, Daillere R, Roberti MP, Yamazaki T, Routy B, et al. Resistance Mechanisms to Immune-

    Checkpoint Blockade in Cancer: Tumor-Intrinsic and -Extrinsic Factors. Immunity 2016;44: 1255-69.

    Research. on June 17, 2021. © 2019 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 27, 2019; DOI: 10.1158/1078-0432.CCR-19-0405

    http://clincancerres.aacrjournals.org/

  • 23

    34. Simon SLabarriere N. PD-1 expression on tumor-specific T cells: Friend or foe for immunotherapy?

    Oncoimmunology 2018;7: e1364828-1-7.

    35. Yokosuka T, Takamatsu M, Kobayashi-Imanishi W, Hashimoto-Tane A, Azuma M, Saito T. Programmed cell death

    1 forms negative costimulatory microclusters that directly inhibit T cell receptor signaling by recruiting

    phosphatase SHP2. J Exp Med 2012;209: 1201-17.

    36. Hui E, Cheung J, Zhu J, Su X, Taylor MJ, Wallweber HA, et al. T cell costimulatory receptor CD28 is a primary

    target for PD-1-mediated inhibition. Science 2017;355: 1428-1433.

    37. Inozume T, Hanada K, Wang QJ, Ahmadzadeh M, Wunderlich JR, Rosenberg SA, et al. Selection of CD8+PD-1+

    lymphocytes in fresh human melanomas enriches for tumor-reactive T cells. J Immunother 2010;33: 956-64.

    38. Gros A, Robbins PF, Yao X, Li YF, Turcotte S, Tran E, et al. PD-1 identifies the patient-specific CD8(+) tumor-

    reactive repertoire infiltrating human tumors. J Clin Invest 2014;124: 2246-59.

    39. Gros A, Parkhurst MR, Tran E, Pasetto A, Robbins PF, Ilyas S, et al. Prospective identification of neoantigen-

    specific lymphocytes in the peripheral blood of melanoma patients. Nat Med 2016;22: 433-8.

    40. Fourcade J, Sun Z, Benallaoua M, Guillaume P, Luescher IF, Sander C, et al. Upregulation of Tim-3 and PD-1

    expression is associated with tumor antigen-specific CD8+ T cell dysfunction in melanoma patients. J Exp Med

    2010;207: 2175-86.

    41. Chauvin JM, Pagliano O, Fourcade J, Sun Z, Wang H, Sander C, et al. TIGIT and PD-1 impair tumor antigen-specific

    CD8(+) T cells in melanoma patients. J Clin Invest 2015;125: 2046-58.

    42. Kamphorst AO, Pillai RN, Yang S, Nasti TH, Akondy RS, Wieland A, et al. Proliferation of PD-1+ CD8 T cells in

    peripheral blood after PD-1-targeted therapy in lung cancer patients. Proc Natl Acad Sci U S A 2017;114: 4993-

    4998.

    43. Wang H, Franco F, Ho PC. Metabolic Regulation of Tregs in Cancer: Opportunities for Immunotherapy. Trends

    Cancer 2017;3: 583-592.

    44. Zhang X, Xiao X, Lan P, Li J, Dou Y, Chen W, et al. OX40 Costimulation Inhibits Foxp3 Expression and Treg

    Induction via BATF3-Dependent and Independent Mechanisms. Cell Rep 2018;24: 607-618.

    Research. on June 17, 2021. © 2019 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 27, 2019; DOI: 10.1158/1078-0432.CCR-19-0405

    http://clincancerres.aacrjournals.org/

  • 24

    45. Long GV, Trefzer U, Davies MA, Kefford RF, Ascierto PA, Chapman PB, et al. Dabrafenib in patients with

    Val600Glu or Val600Lys BRAF-mutant melanoma metastatic to the brain (BREAK-MB): a multicentre, open-label,

    phase 2 trial. Lancet Oncol 2012;13: 1087-95.

    46. Du W, Seah I, Bougazzoul O, Choi G, Meeth K, Bosenberg MW, et al. Stem cell-released oncolytic herpes simplex

    virus has therapeutic efficacy in brain metastatic melanomas. Proc Natl Acad Sci U S A 2017;114: E6157-E6165.

    47. Tiwary S, Morales JE, Kwiatkowski SC, Lang FF, Rao G, McCarty JH. Metastatic Brain Tumors Disrupt the Blood-

    Brain Barrier and Alter Lipid Metabolism by Inhibiting Expression of the Endothelial Cell Fatty Acid Transporter

    Mfsd2a. Sci Rep 2018;8: 8267.

    48. Aspelund A, Antila S, Proulx ST, Karlsen TV, Karaman S, Detmar M, et al. A dural lymphatic vascular system that

    drains brain interstitial fluid and macromolecules. J Exp Med 2015;212: 991-9.

    49. Louveau A, Smirnov I, Keyes TJ, Eccles JD, Rouhani SJ, Peske JD, et al. Structural and functional features of

    central nervous system lymphatic vessels. Nature 2015;523: 337-41.

    50. Blair GE, Dixon SC, Griffiths SA, Zajdel ME. Restricted replication of human adenovirus type 5 in mouse cell lines.

    Virus Res 1989;14: 339-46.

    Research. on June 17, 2021. © 2019 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 27, 2019; DOI: 10.1158/1078-0432.CCR-19-0405

    http://clincancerres.aacrjournals.org/

  • 25

    Figure Legends

    Figure 1. Injection of Delta-24-RGDOX into the primary s.c. melanoma inhibits distant untreated s.c.

    melanoma and induces immune memory. A, A cartoon depiction of the treatment scheme for the schedule

    (left) and position of implantation and viral injection (right). s.c.: subcutaneously; i.t.: intratumorally. B,

    Representative bioluminescent images of mice treated with PBS or Delta-24-RGDOX at indicated time points.

    C, Spider plots of the tumor bioluminescence in the mice from the indicated treatment groups. D, Survival plots

    of the treatment groups (n = 9). E, Survival plots Delta-24-RGDOX treatment survivors after being first re-

    challenged with B16F10 Red-FLuc (left panel, n = 4) and then CMT64 (right panel, n = 4) cells. D24-RGDOX:

    Delta-24-RGDOX; NS: not significant (P ≥ 0.05); *P < 0.01, log-rank test.

    Figure 2. Injection of Delta-24-RGDOX into the s.c. melanoma inhibits distant untreated i.c. melanoma

    and induces immune memory. A, A cartoon depiction of the treatment scheme for the schedule (left) and

    position of implantation and viral injection (right). s.c.: subcutaneously; i.c.: intracranially; i.t.: intratumorally.

    B, Representative bioluminescent images of mice treated with PBS or Delta-24-RGDOX at indicated time

    points. C , Spider plots of the tumor bioluminescence in the mice from the indicated treatment groups. D,

    Survival plots of the treatment groups. PBS: n = 8; D24-RGDOX: n = 11. E, Survival plots for re-challenging

    survivors from Delta-24-RGDOX treatment with B16F10 Red-FLuc-3 (n = 5) cells in the contralateral

    hemisphere. F, Shown are representative hematoxylin-and-eosin-stained, whole-mount coronal sections of the

    mouse brains from Group Naïve (upper panel, euthanized on Day 26) or Survivor (lower panel, euthanized on

    Day 95) from E. Scale bars: 1 mm. D24-RGDOX: Delta-24-RGDOX; NS: not significant (P ≥ 0.05); * P =

    0.005, log-rank test.

    Figure 3. Effect of Delta-24-RGDOX on tumor cells and lymphocytes at the injected tumor. A, Fourteen

    days after s.c. tumor implantation, Delta-24-RGDOX was injected intratumorally. 24 h later, the tumor was

    fixed and processed for immunofluorescent staining for E1A. Representative images are shown. Scale bars: 100

    Research. on June 17, 2021. © 2019 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 27, 2019; DOI: 10.1158/1078-0432.CCR-19-0405

    http://clincancerres.aacrjournals.org/

  • 26

    µm. B, MHC I and MHC II expression in melanoma cells from implanted tumors. After the implantation of

    B16F10-EGFP, Delta-24-RGDOX was injected intratumorally on days 7. After 48 hours, the tumors (taken

    from 3-4 mice/group) were harvested, dissociated, and analyzed with flow cytometry for MHC I and MHC II

    expression. Tumor cells were gated for EGFP+. IgG staining was used as a negative control. The colored

    numbers indicate the MFI for the curve of the same color in the graphs. C-F, The mice with 1 s.c. tumor were

    treated as depicted in Figure 1A. Three days after the last dose of viral injection, tumors were collected (4-5

    mice/group), and the leukocytes in the tumor were enriched and analyzed with flow cytometry to assess the

    number of leukocytes (CD45+) (C); the frequency (D) and number (E) of T cells (CD3+), cytotoxic T cells

    (CD3+ CD8+) and T helper cells (CD3+ CD4+); and the frequency and number of effector T cells (F). D, Left

    panel: Representative flow cytometry plots; the numbers are the frequency of the gated cell populations. Right

    panel: quantification of the cell populations in left panel. D24-RGDOX: Delta-24-RGDOX. Teff: effector T

    cells. Values represent means ± standard deviations (n = 3). *P = 0.03; **P < 0.0002; NS: not significant (P ≥

    0.05); 2-tailed Student t-test.

    Figure 4. Systemic immune activation induced by localized Delta-24-RGDOX treatment in tumor-bearing

    mice. The mice with s.c. and i.c. tumors were treated as depicted in Figure 2A. Three days after the last dose of

    viral injection, tissues were collected from the mice (4-5 mice/group) for immunological analysis. A-D, The

    leukocytes from the blood (A and B) and brain hemispheres with tumor implantation (C and D) were analyzed

    with flow cytometry to assess the frequency of T cells (CD3+), cytotoxic T cells (CD3+ CD8+), and T helper

    cells (CD3+ CD4+) (A and C); and the frequency (B) and number (D) of effector T cells. A and C, Left panel:

    Representative flow cytometry plots; the numbers are the frequency of the gated cell populations. Right panel:

    quantification of the cell populations in left panel. E, Splenocytes and cells from inguinal lymph nodes (tumor-

    draining lymph node of the s.c. tumor) were co-cultured with B16F10 Red-FLuc-3 cells with or without the

    infection of the indicated viruses. The amount of IFNγ secreted into the medium was assessed with ELISA forty

    Research. on June 17, 2021. © 2019 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 27, 2019; DOI: 10.1158/1078-0432.CCR-19-0405

    http://clincancerres.aacrjournals.org/

  • 27

    hours later. D24-RGD: Delta-24-RGD; D24-RGDOX: Delta-24-RGDOX. Teff: effector T cells. Values

    represent means ± standard deviations (n = 3). NS: not significant (P ≥ 0.05); *P < 0.05, 2-tailed Student t-test.

    Figure 5. Disruption of immune suppression in both treated and untreated tumors by Delta-24-RGDOX.

    The mice with s.c. and i.c. tumors were treated as depicted in Figure 2A. Three days after the last dose of viral

    injection, the leukocytes from the virus-injected s.c. tumors, peripheral blood, spleens, inguinal lymph nodes,

    and brain hemispheres with untreated tumor (4 - 5 mice/group) were analyzed with flow cytometry to assess the

    frequency of exhausted T cells (PD-1+ TIM3+) (A) and regulatory T cells (B) and the frequency of PD-1+ cells

    in CD8+ T cells (C). A and B, Upper panel: representative flow cytometry plots; the numbers are the frequency

    of the gated cell populations. Lower panel: quantification of the cell populations in left panel. Values represent

    means ± standard deviations (n = 3). *P < 0.005, 2-tailed Student t-test. D24-RGDOX: Delta-24-RGDOX;

    Treg: regulatory T cell.

    Figure 6. Tumor-specific T-cell expansion and migration to target distant tumors mediated by localized

    Delta-24RGDOX treatment. A, A cartoon depiction of the treatment scheme for the schedule (left) and

    position of implantation and viral injection (right). OVA-specific (OT-I/Luc) T cells (1 106) were injected on

    day 6. s.c.: subcutaneously; i.t.: intratumorally. B, Bioluminescence imaging shows the distribution of tumor-

    specific T cells after i.t. injection of Delta-24-RGDOX in the first implanted s.c. tumor at the indicated times. C,

    Quantification of bioluminescence in the second untreated tumors of the mice shown in B on days 7 and 16. D,

    On day 19, the lymphocytes from the tumors (5 mice/group) were analyzed with flow cytometry to assess

    OVA-Tet+ CD8+ cells. The quantification of this cell population in the tumors is shown. Values represent

    means ± standard deviations (n = 3). Groups A, B, and C in C and D are the same as those indicated in B. E,

    OVA-specific IFNγ-producing cells detected by ELISPOT. After implantation of B16-OVA cells s.c., three

    doses of Delta-24-RGDOX were injected introtumorally on Days 7, 9, 11. On Day 16, splenocytes were isolated

    from the mice (3 mice per group) and stimulated with indicated peptides. IFNγ spot-formaing cells (SFC) were

    Research. on June 17, 2021. © 2019 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 27, 2019; DOI: 10.1158/1078-0432.CCR-19-0405

    http://clincancerres.aacrjournals.org/

  • 28

    measured using an IFNγ ELISPOT assay. D24-RGDOX: Delta-24-RGDOX; NS: not significant (P ≥ 0.05); *P

    < 0.05, 2-tailed Student t-test.

    Research. on June 17, 2021. © 2019 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 27, 2019; DOI: 10.1158/1078-0432.CCR-19-0405

    http://clincancerres.aacrjournals.org/

  • Research. on June 17, 2021. © 2019 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 27, 2019; DOI: 10.1158/1078-0432.CCR-19-0405

    http://clincancerres.aacrjournals.org/

  • Research. on June 17, 2021. © 2019 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 27, 2019; DOI: 10.1158/1078-0432.CCR-19-0405

    http://clincancerres.aacrjournals.org/

  • Research. on June 17, 2021. © 2019 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 27, 2019; DOI: 10.1158/1078-0432.CCR-19-0405

    http://clincancerres.aacrjournals.org/

  • Research. on June 17, 2021. © 2019 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 27, 2019; DOI: 10.1158/1078-0432.CCR-19-0405

    http://clincancerres.aacrjournals.org/

  • Research. on June 17, 2021. © 2019 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 27, 2019; DOI: 10.1158/1078-0432.CCR-19-0405

    http://clincancerres.aacrjournals.org/

  • Research. on June 17, 2021. © 2019 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 27, 2019; DOI: 10.1158/1078-0432.CCR-19-0405

    http://clincancerres.aacrjournals.org/

  • Published OnlineFirst August 27, 2019.Clin Cancer Res Hong Jiang, Dong Ho Shin, Teresa T. Nguyen, et al. Disseminated Subcutaneous and Intracranial MelanomasDelta-24-RGDOX Induces Systemic Immunity against Localized Treatment with Oncolytic Adenovirus

    Updated version

    10.1158/1078-0432.CCR-19-0405doi:

    Access the most recent version of this article at:

    Material

    Supplementary

    http://clincancerres.aacrjournals.org/content/suppl/2019/08/27/1078-0432.CCR-19-0405.DC1

    Access the most recent supplemental material at:

    Manuscript

    Authoredited. Author manuscripts have been peer reviewed and accepted for publication but have not yet been

    E-mail alerts related to this article or journal.Sign up to receive free email-alerts

    Subscriptions

    Reprints and

    [email protected] at

    To order reprints of this article or to subscribe to the journal, contact the AACR Publications

    Permissions

    Rightslink site. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)

    .http://clincancerres.aacrjournals.org/content/early/2019/08/27/1078-0432.CCR-19-0405To request permission to re-use all or part of this article, use this link

    Research. on June 17, 2021. © 2019 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on August 27, 2019; DOI: 10.1158/1078-0432.CCR-19-0405

    http://clincancerres.aacrjournals.org/lookup/doi/10.1158/1078-0432.CCR-19-0405http://clincancerres.aacrjournals.org/content/suppl/2019/08/27/1078-0432.CCR-19-0405.DC1http://clincancerres.aacrjournals.org/cgi/alertsmailto:[email protected]://clincancerres.aacrjournals.org/content/early/2019/08/27/1078-0432.CCR-19-0405http://clincancerres.aacrjournals.org/

    Article FileFigure 1Figure 2Figure 3Figure 4Figure 5Figure 6